Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 348
Filtrer
1.
Food Funct ; 15(17): 8689-8699, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39145543

RÉSUMÉ

Histone deacetylase (HDAC) enzymes play a key role in cell function and are implicated in several diseases such as inflammation, cancer, and neurodegeneration. Studies on natural products have revealed their potential and have led to increased research on natural HDAC inhibitors. Since the progression of these diseases is a prolonged process, dietary supplements and nutraceuticals consisting of plant extracts may be beneficial against HDAC related diseases. Beyond nutritional purposes, cinnamon (Cinnamomum cassia (L.) J. Presl), as a regularly consumed dietary additive due to its rich flavor, may present co-benefits during lifelong use. In this study, cinnamon extracts of differing polarities, trans-cinnamaldehyde and trans-cinnamic acid were evaluated for HDAC 1 inhibitory activity. The total phenol and flavonoid contents were quantified by spectrophotometry, while cinnamaldehyde and cinnamic acid analyses were performed using UPLC-DAD, ESI-MS/MS. Ethanol and dichloromethane extracts yielded the highest cinnamaldehyde and cinnamic acid contents of 389.17 mg per g extract and 11.85 mg per g extract, respectively. The essential oil (IC50: 51.11 µg ml-1) and 70% ethanol extract (IC50: 107.90 µg ml-1) showed the most potent HDAC 1 inhibitory activity. Individually, cinnamaldehyde and cinnamic acid were determined to have IC50 values of 7.58 µg ml-1 and 9.15 µg ml-1, respectively. As the 70% ethanol extract was able to yield remarkably lower cinnamaldehyde and cinnamic acid amounts, the potential of other moderately polar phenolic compounds for HDAC 1 inhibitory activity was revealed. The essential oil and 70% ethanol extracts of Cinnamomum cassia bark can be further evaluated in future studies for use in products against HDAC 1 related diseases.


Sujet(s)
Acroléine , Cinnamates , Cinnamomum zeylanicum , Histone Deacetylase 1 , Inhibiteurs de désacétylase d'histone , Extraits de plantes , Cinnamates/pharmacologie , Cinnamates/analyse , Acroléine/analogues et dérivés , Acroléine/pharmacologie , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Cinnamomum zeylanicum/composition chimique , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 1/métabolisme , Inhibiteurs de désacétylase d'histone/pharmacologie , Humains , Cinnamomum aromaticum/composition chimique
2.
Cell Commun Signal ; 22(1): 361, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39010083

RÉSUMÉ

BACKGROUND: Breast cancer is one of the most lethal cancers in women. Despite significant advances in the diagnosis and treatment of breast cancer, many patients still succumb to this disease, and thus, novel effective treatments are urgently needed. Natural product coumarin has been broadly investigated since it reveals various biological properties in the medicinal field. Accumulating evidence indicates that histone deacetylase inhibitors (HDACIs) are promising novel anti-breast cancer agents. However, most current HDACIs exhibit only moderate effects against solid tumors and are associated with severe side effects. Thus, to develop more effective HDACIs for breast cancer therapy, hydroxamate of HDACIs was linked to coumarin core, and coumarin-hydroxamate hybrids were designed and synthesized. METHODS: A substituted coumarin moiety was incorporated into the classic hydroxamate HDACIs by the pharmacophore fusion strategy. ZN444B was identified by using the HDACI screening kit and cell viability assay. Molecular docking was performed to explore the binding mode of ZN444B with HDAC1. Western blot, immunofluorescent staining, cell viability, colony formation and cell migration and flow cytometry assays were used to analyze the anti-breast cancer effects of ZN444B in vitro. Orthotopic studies in mouse models were applied for preclinical evaluation of efficacy and toxicity in vivo. Proteomic analysis, dual-luciferase reporter assay, chromatin immunoprecipitation, co-immunoprecipitation, immunofluorescent staining assays along with immunohistochemical (IHC) analysis were used to elucidate the molecular basis of the actions of ZN444B. RESULTS: We synthesized and identified a novel coumarin-hydroxamate conjugate, ZN444B which possesses promising anti-breast cancer activity both in vitro and in vivo. A molecular docking model showed that ZN444B binds to HDAC1 with high affinity. Further mechanistic studies revealed that ZN444B specifically decreases FOS-like antigen 2 (FOSL2) mRNA levels by inhibiting the deacetylase activity of HDAC1 on Sp1 at K703 and abrogates the binding ability of Sp1 to the FOSL2 promoter. Furthermore, FOSL2 expression positively correlates with breast cancer progression and metastasis. Silencing FOSL2 expression decreases the sensitivity of breast cancer cells to ZN444B treatment. In addition, ZN444B shows no systemic toxicity in mice. CONCLUSIONS: Our findings highlight the potential of FOSL2 as a new biomarker and therapeutic target for breast cancer and that targeting the HDAC1-Sp1-FOSL2 signaling axis with ZN444B may be a promising therapeutic strategy for breast cancer.


Sujet(s)
Tumeurs du sein , Coumarines , Histone Deacetylase 1 , Acides hydroxamiques , Transduction du signal , Coumarines/composition chimique , Coumarines/pharmacologie , Humains , Histone Deacetylase 1/métabolisme , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 1/génétique , Femelle , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Acides hydroxamiques/pharmacologie , Acides hydroxamiques/composition chimique , Acides hydroxamiques/usage thérapeutique , Facteur de transcription Sp1/métabolisme , Souris , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/composition chimique , Lignée cellulaire tumorale , Simulation de docking moléculaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris nude , Protéines proto-oncogènes c-fos/métabolisme , Protéines proto-oncogènes c-fos/génétique , Souris de lignée BALB C , Mouvement cellulaire/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/usage thérapeutique , Découverte de médicament
3.
Bioorg Chem ; 151: 107556, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39068717

RÉSUMÉ

In recent years, poly(ADP-ribose)polymerase-1 (PARP-1) and histone deacetylase (HDAC) have emerged as significant targets in tumor therapy, garnering widespread attention. In this study, we designed and synthesized two novel phthalazinone PARP-1 inhibitors and dual PARP-1/HDAC-1 inhibitors, named DLC-1-46 containing dithiocarboxylate fragments and DLC-47-63 containing hydroxamic acid fragments, and evaluated their inhibitory activities on enzymes and cells. Among the PARP-1 inhibitors, most compounds exhibited high inhibitory activity against the PARP-1 enzyme, with DLC-1-6 being particularly notable, showing IC50 values <0.2 nM. Notably, DLC-1 demonstrated significant anti-proliferative activity, with IC50 values for inhibiting the proliferation of MDA-MB-436, MDA-MB-231, and MCF-7 cells reaching 0.08, 26.39, and 1.01 µM, respectively. Further investigation revealed that DLC-1 arrested MDA-MB-231 cells in the G1 phase and induced apoptosis in a dose-dependent manner. Among the designed dual PARP-1/HDAC-1 inhibitors, several compounds exhibited potent dual-target inhibitory activity, with DLC-49 displaying IC50 values of 0.53 nM and 17 nM for PARP-1 and HDAC-1, respectively. DLC-50 demonstrated the most potent anti-proliferative activity, with IC50 values for inhibiting the proliferation of MDA-MB-436, MDA-MB-231, and MCF-7 cells at 0.30, 2.70, and 2.41 µM, respectively. Cell cycle arrest and apoptosis assays indicated that DLC-50 arrested the cell cycle in the G2 phase and induced apoptosis in HCT-116 cells. Our findings present a novel avenue for further exploration of PARP-1 inhibitors and dual PARP-1/HDAC-1 inhibitors.


Sujet(s)
Antinéoplasiques , Prolifération cellulaire , Conception de médicament , Tests de criblage d'agents antitumoraux , Histone Deacetylase 1 , Inhibiteurs de désacétylase d'histone , Phtalazines , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 1/métabolisme , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/synthèse chimique , Inhibiteurs de désacétylase d'histone/composition chimique , Relation structure-activité , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Phtalazines/pharmacologie , Phtalazines/synthèse chimique , Phtalazines/composition chimique , Structure moléculaire , Relation dose-effet des médicaments , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Simulation de docking moléculaire
4.
PLoS One ; 19(7): e0306168, 2024.
Article de Anglais | MEDLINE | ID: mdl-39008483

RÉSUMÉ

Dual-targeting chromatin regulation and DNA damage repair signaling presents a promising avenue for cancer therapy. Applying rational drug design, we synthesized a potent dual-targeting small molecule, SP-1-303. Here, we report SP-1-303 as a class I isoform selective histone deacetylase (HDAC) inhibitor and an activator of the ataxia-telangiectasia mutated protein (ATM). In vitro enzymatic assays demonstrated selective inhibition of HDAC1 and HDAC3. Cellular growth inhibition studies show that SP-1-303 differentially inhibits growth of estrogen receptor positive breast cancer (ER+ BC) cells with effective growth inhibition concentrations (EC50) for MCF-7 and T47D cells ranging from 0.32 to 0.34 µM, compared to 1.2-2.5 µM for triple negative breast cancer cells, and ~12 µM for normal breast epithelial cells. Western analysis reveals that SP-1-303 decreases estrogen receptor alpha (ER-α) expression and increases p53 protein expression, while inducing the phosphorylation of ATM and its substrates, BRCA1 and p53, in a time-dependent manner in ER+ BC cells. Pharmacokinetic evaluation demonstrates an area under the curve (AUC) of 5227.55 ng/ml × h with an elimination half-life of 1.26 h following intravenous administration in a rat model. Collectively, SP-1-303 emerges as a novel second generation class I (HDAC1 and HDAC3) selective HDAC inhibitor, and ATM activator, capable of modulating ER expression, and inhibiting growth of ER+ BC cells. Combined targeting of class I HDACs and ATM by SP-1-303 offers a promising therapeutic approach for treating ER+ breast cancers and supports further preclinical evaluation.


Sujet(s)
Protéines mutées dans l'ataxie-télangiectasie , Tumeurs du sein , Prolifération cellulaire , Inhibiteurs de désacétylase d'histone , Humains , Inhibiteurs de désacétylase d'histone/pharmacologie , Protéines mutées dans l'ataxie-télangiectasie/métabolisme , Protéines mutées dans l'ataxie-télangiectasie/antagonistes et inhibiteurs , Femelle , Animaux , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Rats , Prolifération cellulaire/effets des médicaments et des substances chimiques , Histone deacetylases/métabolisme , Lignée cellulaire tumorale , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 1/métabolisme , Cellules MCF-7 , Récepteur alpha des oestrogènes/métabolisme , Récepteurs des oestrogènes/métabolisme
5.
Mar Drugs ; 22(6)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38921561

RÉSUMÉ

Acute myeloid leukemia (AML) is a hematologic malignancy characterized by infiltration of the blood and bone marrow, exhibiting a low remission rate and high recurrence rate. Current research has demonstrated that class I HDAC inhibitors can downregulate anti-apoptotic proteins, leading to apoptosis of AML cells. In the present investigation, we conducted structural modifications of marine cytotoxin Santacruzamate A (SCA), a compound known for its inhibitory activity towards HDACs, resulting in the development of a novel series of potent class I HDACs hydrazide inhibitors. Representative hydrazide-based compound 25c exhibited concentration-dependent induction of apoptosis in AML cells as a single agent. Moreover, 25c exhibited a synergistic anti-AML effect when combined with Venetoclax, a clinical Bcl-2 inhibitor employed in AML therapy. This combination resulted in a more pronounced downregulation of anti-apoptotic proteins Mcl-1 and Bcl-xL, along with a significant upregulation of the pro-apoptotic protein cleaved-caspase3 and the DNA double-strand break biomarker γ-H2AX compared to monotherapy. These results highlighted the potential of 25c as a promising lead compound for AML treatment, particularly when used in combination with Venetoclax.


Sujet(s)
Antinéoplasiques , Apoptose , Composés hétérocycliques bicycliques , Synergie des médicaments , Inhibiteurs de désacétylase d'histone , Leucémie aigüe myéloïde , Sulfonamides , Humains , Sulfonamides/pharmacologie , Sulfonamides/composition chimique , Leucémie aigüe myéloïde/traitement médicamenteux , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/composition chimique , Composés hétérocycliques bicycliques/pharmacologie , Composés hétérocycliques bicycliques/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone deacetylases/métabolisme , Animaux , Caspase-3/métabolisme , Protéine Mcl-1/antagonistes et inhibiteurs
6.
J Biomol Struct Dyn ; 42(11): 5642-5656, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38870352

RÉSUMÉ

Histone deacetylase 1 (HDAC1), a class I HDAC enzyme, is crucial for histone modification. Currently, it is emerged as one of the important biological targets for designing small molecule drugs through cancer epigenetics. Along with synthetic inhibitors different natural inhibitors are showing potential HDAC1 inhibitions. In order to gain insights into the relationship between the molecular structures of the natural inhibitors and HDAC1, different molecular modelling techniques (Bayesian classification, recursive partitioning, molecular docking and molecular dynamics simulations) have been applied on a dataset of 155 HDAC1 nature-inspired inhibitors with diverse scaffolds. The Bayesian study showed acceptable ROC values for both the training set and test sets. The Recursive partitioning study produced decision tree 1 with 6 leaves. Further, molecular docking study was processed for generating the protein ligand complex which identified some potential amino acid residues such as F205, H28, L271, P29, F150, Y204 for the binding interactions in case of natural inhibitors. Stability of these HDAC1-natutal inhibitors complexes has been also evaluated by molecular dynamics simulation study. The current modelling study is an attempt to get a deep insight into the different important structural fingerprints among different natural compounds modulating HDAC1 inhibition.Communicated by Ramaswamy H. Sarma.


Sujet(s)
Découverte de médicament , Épigenèse génétique , Histone Deacetylase 1 , Inhibiteurs de désacétylase d'histone , Simulation de docking moléculaire , Simulation de dynamique moléculaire , Tumeurs , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 1/composition chimique , Histone Deacetylase 1/métabolisme , Inhibiteurs de désacétylase d'histone/composition chimique , Inhibiteurs de désacétylase d'histone/pharmacologie , Découverte de médicament/méthodes , Humains , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Tumeurs/enzymologie , Liaison aux protéines , Produits biologiques/composition chimique , Produits biologiques/pharmacologie , Ligands , Théorème de Bayes , Relation structure-activité , Sites de fixation
7.
J Ocul Pharmacol Ther ; 40(6): 397-406, 2024.
Article de Anglais | MEDLINE | ID: mdl-38608232

RÉSUMÉ

Purpose: Previously, we identified increased retinal degeneration and cytokine response in a mouse model of dry age-related macular degeneration (AMD) in the presence of systemic inflammation from rheumatoid arthritis (RA). Histone deacetylases (HDACs) regulate cytokine production by reducing acetylation and are found to be dysregulated in inflammatory diseases, including RA and AMD. Therefore, this current study investigates the effect of HDAC inhibition on AMD progression in the presence of systemic inflammation. Methods: Collagen induced arthritis (CIA) was induced in C57BL6J mice, followed by sodium iodate (NaIO3)-induced retinal degeneration. Mice were treated with a selective HDAC class I inhibitor, MS-275, and retinal structure [optical coherence tomography (OCT)], function (electroretinography), and molecular changes quantitative real-time polymerase chain reaction (RT-qPCR, Western Blot) were assessed. Results: NaIO3 retinal damage was diminished in CIA mice treated with MS-275 (P ≤ 0.05). While no significant difference was observed in retinal pigment epithelium (RPE) function, a trend in increased c-wave amplitude was detected in CIA + NaIO3 mice treated with MS-275. Finally, we identified decreased Hdac1, Hdac3, and Cxcl9 expression in CIA + NaIO3 mouse RPE/choroid when treated with MS-275 (P ≤ 0.05). Conclusions: Our data demonstrate that HDAC inhibition can reduce the additive effect of NaIO3-induced retinal degeneration in the presence of systemic inflammation by CIA as measured by OCT analysis. In addition, HDAC inhibition in CIA + NaIO3 treated mice resulted in reduced cytokine production. These findings are highly innovative and provide additional support to the therapeutic potential of HDAC inhibitors for dry AMD treatment.


Sujet(s)
Modèles animaux de maladie humaine , Histone Deacetylase 1 , Inhibiteurs de désacétylase d'histone , Inflammation , Iodates , Souris de lignée C57BL , Pyridines , Tomographie par cohérence optique , Animaux , Souris , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/administration et posologie , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Inflammation/traitement médicamenteux , Iodates/administration et posologie , Iodates/toxicité , Pyridines/pharmacologie , Pyridines/administration et posologie , Pyridines/usage thérapeutique , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 1/métabolisme , Benzamides/pharmacologie , Benzamides/administration et posologie , Benzamides/usage thérapeutique , Histone deacetylases/métabolisme , Dégénérescence de la rétine/traitement médicamenteux , Dégénérescence de la rétine/anatomopathologie , Dégénérescence maculaire/traitement médicamenteux , Dégénérescence maculaire/anatomopathologie , Mâle , Électrorétinographie , Composés de pyridinium/pharmacologie , Composés de pyridinium/administration et posologie , Atrophie géographique/traitement médicamenteux
8.
Biomed Pharmacother ; 174: 116537, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38579402

RÉSUMÉ

Chronic Lymphocytic Leukemia (CLL) patients have a defective expression of the proapoptotic protein p66Shc and of its transcriptional factor STAT4, which evoke molecular abnormalities, impairing apoptosis and worsening disease prognosis and severity. p66Shc expression is epigenetically controlled and transcriptionally modulated by STAT4; epigenetic modifiers are deregulated in CLL cells and specific histone deacetylases (HDACs) like HDAC1, are overexpressed. Reactivation of STAT4/p66Shc expression may represent an attractive and challenging strategy to reverse CLL apoptosis defects. New selective class I HDAC inhibitors (HDACis, 6a-g) were developed with increased potency over existing agents and preferentially interfering with the CLL-relevant isoform HDAC1, to unveil the role of class I HDACs in the upregulation of STAT4 expression, which upregulates p66Shc expression and hence normalizes CLL cell apoptosis. 6c (chlopynostat) was identified as a potent HDAC1i with a superior profile over entinostat. 6c induces marked apoptosis of CLL cells compared with SAHA, which was associated with an upregulation of STAT4/p66Shc protein expression. The role of HDAC1, but not HDAC3, in the epigenetic upregulation of STAT4/p66Shc was demonstrated for the first time in CLL cells and was validated in siRNA-induced HDAC1/HDAC3 knock-down EBV-B cells. To sum up, HDAC1 inhibition is necessary to reactivate STAT4/p66Shc expression in patients with CLL. 6c is one of the most potent HDAC1is known to date and represents a novel pharmacological tool for reversing the impairment of the STAT4/p66Shc apoptotic machinery.


Sujet(s)
Apoptose , Lymphocytes B , Inhibiteurs de désacétylase d'histone , Leucémie chronique lymphocytaire à cellules B , Facteur de transcription STAT-4 , Protéine transformante-1 contenant un domaine d'homologie-2 de Src , Humains , Leucémie chronique lymphocytaire à cellules B/traitement médicamenteux , Leucémie chronique lymphocytaire à cellules B/anatomopathologie , Leucémie chronique lymphocytaire à cellules B/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Inhibiteurs de désacétylase d'histone/pharmacologie , Protéine transformante-1 contenant un domaine d'homologie-2 de Src/métabolisme , Protéine transformante-1 contenant un domaine d'homologie-2 de Src/génétique , Facteur de transcription STAT-4/métabolisme , Lymphocytes B/effets des médicaments et des substances chimiques , Lymphocytes B/métabolisme , Histone Deacetylase 1/métabolisme , Histone Deacetylase 1/antagonistes et inhibiteurs , Benzamides/pharmacologie , Mâle , Sujet âgé , Femelle , Adulte d'âge moyen
9.
Cells ; 12(23)2023 11 29.
Article de Anglais | MEDLINE | ID: mdl-38067162

RÉSUMÉ

Abnormal sexual maturity exhibits significant detrimental effects on adult health outcomes, and previous studies have indicated that targeting histone acetylation might serve as a potential therapeutic approach to regulate sexual maturity. However, the mechanisms that account for it remain to be further elucidated. Using the mouse model, we showed that Trichostatin A (TSA), a histone deacetylase (HDAC) inhibitor, downregulated the protein level of Hdac1 in ovaries to promote the apoptosis of granulosa cells (GCs), and thus arrested follicular development and delayed sexual maturity. Using porcine GCs as a cell model, a novel sexual maturity-associated lncRNA, which was named as the stimulatory factor of follicular development (SFFD), transcribed from mitochondrion and mediated by HDAC1, was identified using RNA sequencing. Mechanistically, HDAC1 knockdown significantly reduced the H3K27ac level at the -953/-661 region of SFFD to epigenetically inhibit its transcription. SFFD knockdown released miR-202-3p to reduce the expression of cyclooxygenase 1 (COX1), an essential rate-limited enzyme involved in prostaglandin synthesis. This reduction inhibited the proliferation and secretion of 17ß-estradiol (E2) while promoting the apoptosis of GCs. Consequently, follicular development was arrested and sexual maturity was delayed. Taken together, HDAC1 knockdown-mediated SFFD downregulation promoted the apoptosis of GCs through the miR-202-3p-COX1 axis and lead to delayed sexual maturity. Our findings reveal a novel regulatory network modulated by HDAC1, and HDAC1-mediated SFFD may be a promising new therapeutic target to treat delayed sexual maturity.


Sujet(s)
microARN , ARN long non codant , Maturation sexuelle , Animaux , Femelle , Souris , Apoptose/génétique , Prolifération cellulaire , Cyclooxygenase 1/métabolisme , Cellules de la granulosa/métabolisme , microARN/métabolisme , ARN long non codant/génétique , ARN long non codant/métabolisme , Suidae , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 1/génétique , Histone Deacetylase 1/métabolisme , Acides hydroxamiques/pharmacologie
10.
Cell Death Dis ; 13(4): 324, 2022 04 09.
Article de Anglais | MEDLINE | ID: mdl-35395834

RÉSUMÉ

Histone deacetylases (HDACs) are entwined with the pathogenesis of various cancers and potentially serve as promising therapeutic targets. Herein, we intend to explore the potential role of HDAC1 inhibitor (JSL-1) in the tumorigenesis and metastasis of cholangiocarcinoma (CC) and to highlight the molecular basis of its function. As shown by bioinformatics analysis and immunohistochemical detection, high HDAC1 expression was witnessed in CC tissues relative to matched controls from patients with cholecystitis. The molecular network that HDAC1 silencing reduced the enrichment of HDAC1 and Snail on the TPX2 promoter was identified using immunoprecipitation and chromatin immunoprecipitation assays. Both short hairpin RNA (shRNA)-mediated knockdown of HDAC1 and JSL-1 treatment exhibited anti-proliferative, anti-migration and anti-invasion effects on CC cells through downregulation of TPX2. The in vivo xenograft model was developed in nude mice. Consistently, the anti-tumorigenic and anti-metastatic properties of shRNA against HDAC1 and HDAC1 inhibitor were validated in the in vivo settings. Taken together, our data supported the notion that HDAC1 inhibitor retards the initiation and development of CC via mediating the TPX2/Snail axis, highlighting the anti-tumor molecular network functioned in CC.


Sujet(s)
Cholangiocarcinome , Histone Deacetylase 1 , Inhibiteurs de désacétylase d'histone , Animaux , Protéines du cycle cellulaire/génétique , Lignée cellulaire tumorale , Cholangiocarcinome/génétique , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 1/génétique , Inhibiteurs de désacétylase d'histone/pharmacologie , Humains , Souris , Souris nude , Protéines associées aux microtubules/génétique , Métastase tumorale , Petit ARN interférent , Facteurs de transcription de la famille Snail/génétique
11.
BMC Pulm Med ; 22(1): 61, 2022 Feb 11.
Article de Anglais | MEDLINE | ID: mdl-35148729

RÉSUMÉ

BACKGROUND: Exposure to toluene diisocyanate (TDI) is a significant pathogenic factor for asthma. We previously reported that the receptor for advanced glycation end products (RAGE) plays a key role in TDI-induced asthma. Histone deacetylase (HDAC) has been reported to be important in asthmatic pathogenesis. However, its effect on TDI-induced asthma is not known. The aim of this study was to determine the role of RAGE and HDAC in regulating airway inflammation using a TDI-induced murine asthma model. METHODS: BALB/c mice were sensitized and challenged with TDI to establish an asthma model. FPS-ZM1 (RAGE inhibitor), JNJ-26482585 and romidepsin (HDAC inhibitors) were administered intraperitoneally before each challenge. In vitro, the human bronchial epithelial cell line 16HBE was stimulated with TDI-human serum albumin (TDI-HSA). RAGE knockdown cells were constructed and evaluated, and MK2006 (AKT inhibitor) was also used in the experiments. RESULTS: In TDI-induced asthmatic mice, the expression of RAGE, HDAC1, and p-AKT/t-AKT was upregulated, and these expressions were attenuated by FPS-ZM1. Airway reactivity, Th2 cytokine levels in lymph supernatant, IgE, airway inflammation, and goblet cell metaplasia were significantly increased in the TDI-induced asthmatic mice. These increases were suppressed by JNJ-26482585 and romidepsin. In addition, JNJ-26482585 and romidepsin ameliorated the redistribution of E-cadherin and ß-catenin in TDI-induced asthma. In TDI-HSA-stimulated 16HBE cells, knockdown of RAGE attenuated the upregulation of HDAC1 and phospho-AKT (p-AKT). Treatment with the AKT inhibitor MK2006 suppressed TDI-induced HDAC1 expression. CONCLUSIONS: These findings indicate that RAGE modulates HDAC1 expression via the PI3K/AKT pathway, and that inhibition of HDAC prevents TDI-induced airway inflammation.


Sujet(s)
Asthme/prévention et contrôle , Histone Deacetylase 1/métabolisme , Inflammation/prévention et contrôle , Récepteur spécifique des produits finaux de glycosylation avancée/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Asthme/induit chimiquement , Benzamides/pharmacologie , Lignée cellulaire , Cytokines/métabolisme , Depsipeptides/pharmacologie , Modèles animaux de maladie humaine , Histone Deacetylase 1/antagonistes et inhibiteurs , Humains , Mâle , Souris , Souris de lignée BALB C , Phosphatidylinositol 3-kinases/métabolisme , Récepteur spécifique des produits finaux de glycosylation avancée/antagonistes et inhibiteurs , 2,4-Diisocyanato-1-méthyl-benzène/toxicité
12.
J Invest Dermatol ; 142(1): 77-87.e10, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34284046

RÉSUMÉ

HDAC inhibitors show therapeutic promise for skin malignancies; however, the roles of specific HDACs in adult epidermal homeostasis and in disease are poorly understood. We find that homozygous epidermal codeletion of Hdac1 and Hdac2 in adult mouse epidermis causes reduced basal cell proliferation, apoptosis, inappropriate differentiation, and eventual loss of Hdac1/2-null keratinocytes. Hdac1/2-deficient epidermis displays elevated acetylated p53 and increased expression of the senescence gene p16. Loss of p53 partially restores basal proliferation, whereas p16 deletion promotes long-term survival of Hdac1/2-null keratinocytes. In activated GLI2-driven pre-basal cell carcinoma, Hdac1/2 deletion dramatically reduces proliferation and increases apoptosis, and knockout of either p53 or p16 partially rescues both proliferation and basal cell viability. Topical application of the HDAC inhibitor romidepsin to the normal epidermis or to GLI2ΔN-driven lesions produces similar defects to those caused by genetic Hdac1/2 deletion, and these are partially rescued by loss of p16. These data reveal essential roles for HDAC1/2 in maintaining proliferation and survival of adult epidermal and basal cell carcinoma progenitors and suggest that the efficacy of therapeutic HDAC1/2 inhibition will depend in part on the mutational status of p53 and p16.


Sujet(s)
Carcinome basocellulaire/métabolisme , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Épiderme/physiologie , Kératinocytes/physiologie , Tumeurs cutanées/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Animaux , Antibiotiques antinéoplasiques/pharmacologie , Antibiotiques antinéoplasiques/usage thérapeutique , Apoptose , Carcinogenèse , Carcinome basocellulaire/traitement médicamenteux , Carcinome basocellulaire/génétique , Différenciation cellulaire , Lignée cellulaire tumorale , Prolifération cellulaire , Inhibiteur p16 de kinase cycline-dépendante/génétique , Depsipeptides/pharmacologie , Depsipeptides/usage thérapeutique , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 1/génétique , Histone Deacetylase 2/antagonistes et inhibiteurs , Histone Deacetylase 2/génétique , Humains , Souris , Souris de lignée C57BL , Souris knockout , États précancéreux , Tumeurs cutanées/traitement médicamenteux , Tumeurs cutanées/génétique , Protéine p53 suppresseur de tumeur/génétique
13.
Eur J Med Chem ; 229: 114049, 2022 Feb 05.
Article de Anglais | MEDLINE | ID: mdl-34954594

RÉSUMÉ

Histone deacetylases (HDAC) are clinically validated and attractive epigenetic drug targets for human cancers. Several HDAC inhibitors have been approved for cancer treatment to date, however, clinical applications have been limited due to the poor pharmacokinetics, bioavailability, selectivity of the HDAC inhibitors and most of them need to be combined with other drugs to achieve better results. Here, we describe our efforts toward the discovery of a novel series of lactam-based derivatives as selective HDAC inhibitors. Intensive structural modifications lead to the identification of compound 24g as the most active Class I HDAC Inhibitor, along with satisfactory metabolic stability in vitro (t1/2, human = 797 min) and the desirable oral bioavailability (F = 92%). More importantly, compound 24g showed good antitumor efficacy in a TMD-8 xenograft model (TGI = 77%) without obvious toxicity. These results indicated that Class I HDAC Inhibitor could be potentially used to treat certain diffuse large B-cell lymphoma therapeutics.


Sujet(s)
Conception de médicament , Histone Deacetylase 1/antagonistes et inhibiteurs , Inhibiteurs de désacétylase d'histone/composition chimique , Animaux , Sites de fixation , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Chiens , Tests de criblage d'agents antitumoraux , Période , Histone Deacetylase 1/métabolisme , Inhibiteurs de désacétylase d'histone/métabolisme , Inhibiteurs de désacétylase d'histone/pharmacocinétique , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Humains , Lymphome B diffus à grandes cellules/traitement médicamenteux , Souris , Microsomes du foie/métabolisme , Simulation de docking moléculaire , Isoformes de protéines/antagonistes et inhibiteurs , Isoformes de protéines/métabolisme , Rats , Relation structure-activité
14.
Fertil Steril ; 117(2): 433-443, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34809976

RÉSUMÉ

OBJECTIVE: To evaluate the effect of inhibition of histone deacetylases (HDACs) by suberoylanilide hydroxamic acid (SAHA) treatment of human uterine leiomyoma primary (HULP) cells in vitro on cell proliferation, cell cycle, extracellular matrix (ECM) formation, and transforming growth factor ß3 (TGF-ß3) signaling. DESIGN: Prospective study comparing uterine leiomyoma (UL) vs. adjacent myometrium (MM) tissue and cells with or without SAHA treatment. SETTING: Hospital and university laboratories. PATIENT(S): Women with UL without any hormone treatment. INTERVENTION(S): Myomectomy or hysterectomy surgery in women for leiomyoma disease. MAIN OUTCOME MEASURE(S): HDAC activity was assessed by enzyme-linked immunosorbent assay, and gene expression was assessed by quantitative real-time polymerase chain reaction. Effects of SAHA on HULP cells were analyzed by CellTiter (Promega, Madison, Wisconsin), Western blot, and quantitative real-time polymerase chain reaction. RESULT(S): The expression of HDAC genes (HDAC1, fold change [FC] = 1.65; HDAC3, FC = 2.08; HDAC6, FC = 2.42) and activity (0.56 vs. 0.10 optical density [OD]/h/mg) was significantly increased in UL vs. MM tissue. SAHA decreased HDAC activity in HULP cells but not in MM cells. Cell viability significantly decreased in HULP cells (81.68% at 5 µM SAHA, 73.46% at 10 µM SAHA), but not in MM cells. Proliferating cell nuclear antigen expression was significantly inhibited in SAHA-treated HULP cells (5 µM SAHA, FC = 0.556; 10 µM SAHA, FC = 0.622). Cell cycle markers, including C-MYC (5 µM SAHA, FC = 0.828) and CCND1 (5 µM SAHA, FC = 0.583; 10 µM SAHA, FC = 0.482), were significantly down-regulated after SAHA treatment. SAHA significantly inhibited ECM protein expression, including FIBRONECTIN (5 µM SAHA, FC = 0.815; 10 µM SAHA, FC = 0.673) and COLLAGEN I (5 µM SAHA, FC = 0.599; 10 µM SAHA, FC = 0.635), in HULP cells. TGFß3 and MMP9 gene expression was also significantly down-regulated by 10 µM SAHA (TGFß3, FC = 0.596; MMP9, FC = 0.677). CONCLUSION(S): SAHA treatment inhibits cell proliferation, cell cycle, ECM formation, and TGF-ß3 signaling in HULP cells, suggesting that histone deacetylation may be useful for treatment of UL.


Sujet(s)
Antinéoplasiques/pharmacologie , Inhibiteurs de désacétylase d'histone/pharmacologie , Léiomyome/traitement médicamenteux , Tumeurs de l'utérus/traitement médicamenteux , Vorinostat/pharmacologie , Adulte , Cycle cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Matrice extracellulaire/effets des médicaments et des substances chimiques , Matrice extracellulaire/métabolisme , Matrice extracellulaire/anatomopathologie , Femelle , Régulation de l'expression des gènes tumoraux , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 1/génétique , Histone Deacetylase 1/métabolisme , Histone deacetylase 6/antagonistes et inhibiteurs , Histone deacetylase 6/génétique , Histone deacetylase 6/métabolisme , Histone deacetylases/génétique , Histone deacetylases/métabolisme , Humains , Léiomyome/enzymologie , Léiomyome/génétique , Léiomyome/anatomopathologie , Adulte d'âge moyen , Études prospectives , Transduction du signal , Facteur de croissance transformant bêta-3/métabolisme , Cellules cancéreuses en culture , Tumeurs de l'utérus/enzymologie , Tumeurs de l'utérus/génétique , Tumeurs de l'utérus/anatomopathologie
15.
Eur J Med Chem ; 227: 113893, 2022 Jan 05.
Article de Anglais | MEDLINE | ID: mdl-34656899

RÉSUMÉ

The equilibrium between histone acetylation and deacetylation plays an important role in cancer initiation and progression. The histone deacetylases (HDACs) are a class of key regulators of gene expression that enzymatically remove an acetyl moiety from acetylated lysine ε-amino groups on histone tails. Therefore, HDAC inhibitors have recently emerged as a promising strategy for cancer therapy and several pan-HDAC inhibitors have globally been approved for clinical use. In the present study, we designed and synthesized a series of substituted indole-based hydroxamic acid derivatives that exhibited potent anti-proliferative activities in various tumor cell lines. Among the compounds tested, compound 4o, was found to be among the most potent in the inhibition of HDAC1 (half maximal inhibitory concentration, IC50 = 1.16 nM) and HDAC6 (IC50 = 2.30 nM). It also exhibited excellent in vitro anti-tumor proliferation activity. Additionally, compound 4o effectively increased the acetylation of histone H3 in a dose-dependent manner and inhibited cell proliferation by inducing cell cycle arrest and apoptosis. Moreover, compound 4o remarkably blocked colony formation in HCT116 cancer cells. Based on its favorable in vitro profile, compound 4o was further evaluated in an HCT116 xenograft mouse model, in which it demonstrated better in vivo efficacy than the clinically used HDAC inhibitor, suberanilohydroxamic acid. Interestingly, compound 4k was found to have a preference for the inhibition of HDAC6, with IC50 values of 115.20 and 5.29 nM against HDAC1 and HDAC6, respectively.


Sujet(s)
Antinéoplasiques/pharmacologie , Conception de médicament , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone deacetylase 6/antagonistes et inhibiteurs , Inhibiteurs de désacétylase d'histone/pharmacologie , Acides hydroxamiques/pharmacologie , Indoles/pharmacologie , Animaux , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Tests de criblage d'agents antitumoraux , Histone Deacetylase 1/métabolisme , Histone deacetylase 6/métabolisme , Inhibiteurs de désacétylase d'histone/synthèse chimique , Inhibiteurs de désacétylase d'histone/composition chimique , Humains , Acides hydroxamiques/synthèse chimique , Acides hydroxamiques/composition chimique , Indoles/composition chimique , Mâle , Souris , Souris de lignée BALB C , Souris nude , Structure moléculaire , Tumeurs expérimentales/traitement médicamenteux , Tumeurs expérimentales/métabolisme , Tumeurs expérimentales/anatomopathologie , Relation structure-activité , Cellules cancéreuses en culture
16.
Eur J Med Chem ; 227: 113908, 2022 Jan 05.
Article de Anglais | MEDLINE | ID: mdl-34656900

RÉSUMÉ

Highly efficacious and tolerable agents for the treatment of glioblastoma (GBM), the most common and aggressive primary brain tumor, are urgently needed. Herein, we reveal the design, synthesis and biological evaluation of several piperazine based benzamide derivatives, which are based on the non-classical isostere principle and combination principle for GBM therapy. After structure-activity relationship (SAR) study, compound L19 was demonstrated as the most promising compound with IC50 values of 0.15 µM, 0.29 µM, 1.25 µM against GBM C6, U87-MG, U251 cells, respectively. Moreover, compound L19 could inhibit the proliferation, migration and invasion, as well as induce apoptosis and cell cycle arrest of GBM cell lines in vitro. From mechanism perspective, compound L19 could regulate the cell cycle-related proteins and influence the p16INK4a-CDK4/6-pRb pathway by western blotting experiment. What is worth mentioning is that compound L19 could penetrate the blood-brain barrier (BBB) with an exceptional brain-to-plasma ratio of 1.07 in vivo. Besides, the superior anti-glioblastoma potency in vivo of compound L19 was identified on U87-MG-xenograft model without any apparent host toxicity. Overall, the potential of compound L19 warrants further pre-clinical investigation for GBM therapy.


Sujet(s)
Antinéoplasiques/pharmacologie , Benzamides/pharmacologie , Glioblastome/traitement médicamenteux , Inhibiteurs de désacétylase d'histone/pharmacologie , Pipérazines/pharmacologie , Animaux , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Benzamides/synthèse chimique , Benzamides/composition chimique , Cycle cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Tests de criblage d'agents antitumoraux , Femelle , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 1/métabolisme , Inhibiteurs de désacétylase d'histone/synthèse chimique , Inhibiteurs de désacétylase d'histone/composition chimique , Humains , Mâle , Souris , Lignées consanguines de souris , Souris nude , Structure moléculaire , Tumeurs expérimentales/traitement médicamenteux , Tumeurs expérimentales/métabolisme , Tumeurs expérimentales/anatomopathologie , Pipérazines/composition chimique , Rats , Rat Sprague-Dawley , Relation structure-activité , Cellules cancéreuses en culture
17.
Mediators Inflamm ; 2021: 6359652, 2021.
Article de Anglais | MEDLINE | ID: mdl-34924813

RÉSUMÉ

Ellagic acid (EA) was reported to play protective roles in rheumatoid arthritis (RA). It was found that the level of metastasis-associated gene 1 (MTA1)/histone deacetylase 1 (HDAC1) protein complex was downregulated by polyphenols in several human disorders. Notably, inhibition of MTA1 or HDAC1 has anti-inflammatory effects on RA. Therefore, our study is aimed at investigating whether EA prevents RA progression through regulating the MTA1/HDAC1 complex. Herein, the human fibroblast-like synoviocyte (FLS) cell line MH7A was treated with TNF-α to induce an inflammation model in vitro and then incubated with different concentrations of EA. Western blot analysis showed that EA reduced MTA1 expression in a dose-dependent manner in MH7A cells. Then, TNF-α-treated MH7A cells were incubated with EA alone or together with MTA1 overexpression plasmid (pcDNA-MTA1), and we found that EA inhibited proliferation, inflammation cytokine levels, and oxidative stress marker protein levels and promoted apoptosis in MH7A cells, while MTA1 overexpression abolished these effects. Moreover, coimmunoprecipitation assay verified the interaction between MTA1 and HDAC1. EA downregulated the MTA1/HDAC1 complex in MH7A cells. MTA1 knockdown inhibited proliferation, inflammation, and oxidative stress and promoted apoptosis in MH7A cells, while HDAC1 overexpression reversed these effects. Moreover, chromatin immunoprecipitation assay indicated that EA inhibited HDAC1-mediated Nur77 deacetylation. Rescue experiments demonstrated that Nur77 knockdown reversed the effects of EA on MH7A cell biological behaviors. Additionally, EA treatment attenuated arthritis index, paw swelling, synovial hyperplasia, and inflammation in collagen-induced arthritis (CIA) rats. In conclusion, EA inhibited proliferation, inflammation, and oxidative stress and promoted apoptosis in MH7A cells and alleviated the severity of RA in CIA rats though downregulating MTA1/HDAC1 complex and promoting HDAC1 deacetylation-mediated Nur77 expression.


Sujet(s)
Polyarthrite rhumatoïde/traitement médicamenteux , Acide ellagique/pharmacologie , Histone Deacetylase 1/antagonistes et inhibiteurs , Protéines de répression/antagonistes et inhibiteurs , Transactivateurs/antagonistes et inhibiteurs , Acétylation , Animaux , Apoptose/effets des médicaments et des substances chimiques , Cellules cultivées , Histone Deacetylase 1/physiologie , Humains , Mâle , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/physiologie , Stress oxydatif/effets des médicaments et des substances chimiques , Rats , Rat Wistar , Protéines de répression/physiologie , Transactivateurs/physiologie
18.
Bioorg Chem ; 117: 105407, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34653945

RÉSUMÉ

A series of quinazolinyl-containing benzamide derivatives were designed, synthesized and evaluated for their in vitro histone deacetylase 1 (HDAC1) inhibitory activities. Compounds 11a surpassed the known class I selective HDAC inhibitor MS-275 in both HDAC1 enzymatic inhibitory activity and cellular anti-proliferative activity against a selected set of cancer cell types (Hut78, K562, Hep3B and HCT116 cells) with no observed effects on human normal cells. In particular, compound 11a inhibited HDAC1 over the other tested HDACs isoforms (HDAC2, HDAC6 and HDAC8) with acceptable safety profiles. Moreover, compound 11a displayed favorable oral pharmacokinetic properties and showed significant antitumor activity in the A549 tumor xenograft model in vivo.


Sujet(s)
Antinéoplasiques/pharmacologie , Benzamides/pharmacologie , Histone Deacetylase 1/antagonistes et inhibiteurs , Inhibiteurs de désacétylase d'histone/pharmacologie , Animaux , Antinéoplasiques/composition chimique , Antinéoplasiques/usage thérapeutique , Apoptose/effets des médicaments et des substances chimiques , Benzamides/composition chimique , Benzamides/usage thérapeutique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tests de criblage d'agents antitumoraux , Histone Deacetylase 1/métabolisme , Inhibiteurs de désacétylase d'histone/composition chimique , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Humains , Souris de lignée ICR , Simulation de docking moléculaire
19.
Reprod Sci ; 28(12): 3540-3546, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34668144

RÉSUMÉ

Breast cancer resistance protein (BCRP/ABCG2) is a critical drug efflux transporters by limiting drugs' transplacental transfer rates. More investigations on the regulation of placental BCRP offer great promise for enabling pronounced progress in individualized and safe pharmacotherapy during pregnancy. Histone deacetylases (HDACs) play an important role in epigenetic regulation of placental genes. It was reported recently by us that HDAC1 was involved in placental BCRP regulation in vitro. The aim of this study was to further explore the effect of HDAC1 on placental BCRP expression and functionality in animals. Randomly assigned C57BL pregnant dams received intraperitoneal injections of a negative control siRNA or Hdac1 siRNA from embryonic day 7.5 (E7.5) to E15.5, respectively. At E16.5, glyburide (GLB), a probe for evaluating placental BCRP efflux functionality, was injected via the tail vein. Animals were sacrificed through cervical dislocation at various times (5-180 min) after drug administration. The maternal blood, placentas, and fetal-units were collected. GLB concentrations were determined by a validated high-performance liquid chromatography/mass spectrometry (HPLC-MS) assay. Real-time quantitative PCR (qRT-PCR), Western blot, and immunohistochemical (IHC) analysis were employed to identify mRNA/protein levels and localization of gene expressions, respectively. It was noted that Hdac1 inhibition significantly decreased placental Bcrp expression, with markedly increases of GLB concentrations and area under the concentration-time curve (AUC) in fetal-units. Particularly, the ratios of fetal-unit/maternal plasma GLB concentrations were also significantly elevated following Hdac1 repression. Taken together, these findings suggested that HDAC1 was involved in positive regulation of placental BCRP expression and functionality in vivo.


Sujet(s)
Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/biosynthèse , Histone Deacetylase 1/biosynthèse , Préparations pharmaceutiques/administration et posologie , Préparations pharmaceutiques/métabolisme , Placenta/métabolisme , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/antagonistes et inhibiteurs , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/génétique , Animaux , Femelle , Extinction de l'expression des gènes/effets des médicaments et des substances chimiques , Extinction de l'expression des gènes/physiologie , Glibenclamide/administration et posologie , Glibenclamide/métabolisme , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 1/génétique , Souris , Souris de lignée C57BL , Placenta/effets des médicaments et des substances chimiques , Grossesse , Petit ARN interférent/administration et posologie , Petit ARN interférent/métabolisme
20.
Respir Res ; 22(1): 239, 2021 Aug 31.
Article de Anglais | MEDLINE | ID: mdl-34465322

RÉSUMÉ

BACKGROUND: It has been found that up-regulation of histone deacetylases 1 (HDAC1) is involved in the development of pulmonary arterial hypertension (PAH). However, it is still unclear whether inhibition of HDAC1 suppresses the development of PAH via restoring miR-34a level in monocrotaline (MCT)-induced PAH rats. METHODS: PAH rat models were induced by intraperitoneal injection of MCT. HDAC1 was suppressed by intraperitoneal injection of the class I HDAC inhibitor MS-275, and miR-34a was over-expressed via tail vein injection of miR-34a agomiR. RESULTS: HDAC1 protein was significantly increased in MCT-induced PAH rats; this was accompanied with down-regulation of miR-34a and subsequent up-regulation of matrix metalloproteinase 9 (MMP-9)/tissue inhibitor of metalloproteinase 1 (TIMP-1) and MMP-2/TIMP-2. Administration of PAH rats with MS-275 or miR-34a agomiR dramatically abolished MCT-induced reduction of miR-34a and subsequent up-regulation of MMP-9/TIMP-1 and MMP-2/TIMP-2, finally reduced extracellular matrix (ECM) accumulation, pulmonary arterial remodeling, right ventricular systolic pressure (RVSP) and right ventricle hypertrophy index (RVHI) in PAH rats. CONCLUSIONS: HDAC1 contributes to the development of MCT-induced rat PAH by suppressing miR-34a level and subsequently up-regulating the ratio of MMP-9/TIMP-1 and MMP-2/TIMP-2. Inhibition of HDAC1 alleviates pulmonary arterial remodeling and PAH through up-regulation of miR-34a level and subsequent reduction of MMP-9/TIMP-1 and MMP-2/TIMP-2, suggesting that inhibition of HDAC1 might have potential value in the management of PAH.


Sujet(s)
Histone Deacetylase 1/métabolisme , Inhibiteurs de désacétylase d'histone/pharmacologie , Monocrotaline/toxicité , Artère pulmonaire/métabolisme , Remodelage vasculaire/physiologie , Animaux , Benzamides/pharmacologie , Benzamides/usage thérapeutique , Histone Deacetylase 1/antagonistes et inhibiteurs , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Mâle , microARN/biosynthèse , Hypertension artérielle pulmonaire/induit chimiquement , Hypertension artérielle pulmonaire/traitement médicamenteux , Hypertension artérielle pulmonaire/métabolisme , Artère pulmonaire/effets des médicaments et des substances chimiques , Pyridines/pharmacologie , Pyridines/usage thérapeutique , Rats , Rat Sprague-Dawley , Régulation positive/effets des médicaments et des substances chimiques , Régulation positive/physiologie , Remodelage vasculaire/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE